Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters










Publication year range
1.
J Med Chem ; 65(2): 1265-1282, 2022 01 27.
Article in English | MEDLINE | ID: mdl-35081715

ABSTRACT

NIMA-related kinase 1 (Nek1) has lately garnered attention for its widespread function in ciliogenesis, apoptosis, and the DNA-damage response. Despite its involvement in various diseases and its potential as a cancer drug target, no directed medicinal chemistry efforts toward inhibitors against this dark kinase are published. Here, we report the structure-guided design of a potent small-molecule Nek1 inhibitor, starting from a scaffold identified by kinase cross-screening analysis. Seven lead compounds were identified in silico and evaluated for their inhibitory activity. The top compound, 10f, was further profiled for efficacy, toxicity, and bioavailability in a zebrafish polycystic kidney disease model. Administration of 10f caused the expansion of fluorescence-labeled proximal convoluted tubules, supporting our hypothesis that Nek1-inhibition causes cystic kidneys in zebrafish embryos. Compound 10f displayed insignificant inhibition in 48 of 50 kinases in a selectivity test panel. The findings provide a powerful tool to further elucidate the function and pharmacology of this neglected kinase.


Subject(s)
Drug Design , Embryo, Nonmammalian/drug effects , NIMA-Related Kinase 1/antagonists & inhibitors , Polycystic Kidney Diseases/drug therapy , Pronephros/drug effects , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Animals , Embryo, Nonmammalian/enzymology , Polycystic Kidney Diseases/enzymology , Polycystic Kidney Diseases/pathology , Pronephros/embryology , Pronephros/enzymology , Zebrafish
2.
Dev Biol ; 481: 160-171, 2022 01.
Article in English | MEDLINE | ID: mdl-34666023

ABSTRACT

The corpuscles of Stannius (CS) represent a unique endocrine organ of teleostean fish that secrets stanniocalcin-1 (Stc1) to maintain calcium homeostasis. Appearing at 20-25 somite stage in the distal zebrafish pronephros, stc1-expressing cells undergo apical constriction, and are subsequently extruded to form a distinct gland on top of the distal pronephric tubules at 50 â€‹h post fertilization (hpf). Several transcription factors (e.g. Hnf1b, Irx3b, Tbx2a/b) and signaling pathways (e.g. Notch) control CS development. We report now that Fgf signaling is required to commit tubular epithelial cells to differentiate into stc1-expressing CS cells. Inhibition of Fgf signaling by SU5402, dominant-negative Fgfr1, or depletion of fgf8a prevented CS formation and stc1 expression. Ablation experiments revealed that CS have the ability to partially regenerate via active cell migration involving extensive filopodia and lamellipodia formation. Activation of Wnt signaling curtailed stc1 expression, but had no effect on CS formation. Thus, our observations identify Fgf signaling as a crucial component of CS cell fate commitment.


Subject(s)
Cell Differentiation , Endocrine Glands/embryology , Fibroblast Growth Factors , Pronephros/embryology , Wnt Signaling Pathway , Zebrafish Proteins , Zebrafish , Animals , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
3.
Am J Physiol Renal Physiol ; 320(5): F826-F837, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33749326

ABSTRACT

Developing organisms need to adapt to environmental variations as well as to rapid changes in substrate availability and energy demands imposed by fast-growing tissues and organs. Little is known about the adjustments that kidneys undergo in response to these challenges. We performed single-cell RNA sequencing of zebrafish pronephric duct cells to understand how the developing kidney responds to changes in filtered substrates and intrinsic energy requirements. We found high levels of glucose transporters early in development and increased expression of monocarboxylate transporters at later times. This indicates that the zebrafish embryonic kidney displays a high glucose transporting capacity during early development, which is replaced by the ability to absorb monocarboxylates and amino acids at later stages. This change in transport capacity was accompanied by the upregulation of mitochondrial carriers, indicating a switch to increased oxidative phosphorylation to meet the increasing energy demand of a developing kidney.NEW & NOTEWORTHY The zebrafish embryonic kidney has high levels of glucose transporters during early development, which are replaced by monocarboxylate and amino acid transporters later on. Inhibition of Na+-glucose cotransporter-dependent glucose transport by sotagliflozin also increased slc2a1a expression, supporting the idea that the glucose transport capacity is dynamically adjusted during zebrafish pronephros development. Concurrent upregulation of mitochondrial SCL25 transporters at later stages supports the idea that the pronephros adjusts to changing substrate supplies and/or energy demands during embryonic development.


Subject(s)
Energy Metabolism/genetics , Gene Expression Profiling , Pronephros/metabolism , RNA, Messenger/genetics , Single-Cell Analysis , Solute Carrier Proteins/genetics , Transcriptome , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Gene Expression Regulation, Developmental , Pronephros/embryology , RNA, Messenger/metabolism , RNA-Seq , Solute Carrier Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/metabolism
4.
J Am Soc Nephrol ; 32(3): 580-596, 2021 03.
Article in English | MEDLINE | ID: mdl-33593823

ABSTRACT

BACKGROUND: Galloway-Mowat syndrome (GAMOS) is characterized by neurodevelopmental defects and a progressive nephropathy, which typically manifests as steroid-resistant nephrotic syndrome. The prognosis of GAMOS is poor, and the majority of children progress to renal failure. The discovery of monogenic causes of GAMOS has uncovered molecular pathways involved in the pathogenesis of disease. METHODS: Homozygosity mapping, whole-exome sequencing, and linkage analysis were used to identify mutations in four families with a GAMOS-like phenotype, and high-throughput PCR technology was applied to 91 individuals with GAMOS and 816 individuals with isolated nephrotic syndrome. In vitro and in vivo studies determined the functional significance of the mutations identified. RESULTS: Three biallelic variants of the transcriptional regulator PRDM15 were detected in six families with proteinuric kidney disease. Four families with a variant in the protein's zinc-finger (ZNF) domain have additional GAMOS-like features, including brain anomalies, cardiac defects, and skeletal defects. All variants destabilize the PRDM15 protein, and the ZNF variant additionally interferes with transcriptional activation. Morpholino oligonucleotide-mediated knockdown of Prdm15 in Xenopus embryos disrupted pronephric development. Human wild-type PRDM15 RNA rescued the disruption, but the three PRDM15 variants did not. Finally, CRISPR-mediated knockout of PRDM15 in human podocytes led to dysregulation of several renal developmental genes. CONCLUSIONS: Variants in PRDM15 can cause either isolated nephrotic syndrome or a GAMOS-type syndrome on an allelic basis. PRDM15 regulates multiple developmental kidney genes, and is likely to play an essential role in renal development in humans.


Subject(s)
DNA-Binding Proteins/genetics , Hernia, Hiatal/genetics , Microcephaly/genetics , Mutation, Missense , Nephrosis/genetics , Transcription Factors/genetics , Amino Acid Sequence , Amino Acid Substitution , Animals , Cell Line , Child, Preschool , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/deficiency , Female , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Gene Knockout Techniques , High-Throughput Nucleotide Sequencing , Humans , Infant , Infant, Newborn , Male , Models, Molecular , Nephrotic Syndrome/genetics , Podocytes/metabolism , Polymorphism, Single Nucleotide , Pronephros/embryology , Pronephros/metabolism , Protein Stability , Transcription Factors/chemistry , Transcription Factors/deficiency , Xenopus laevis/embryology , Xenopus laevis/genetics , Zinc Fingers/genetics
5.
Cells ; 9(5)2020 05 20.
Article in English | MEDLINE | ID: mdl-32443839

ABSTRACT

Automated high-throughput workflows allow for chemical toxicity testing and drug discovery in zebrafish disease models. Due to its conserved structural and functional properties, the zebrafish pronephros offers a unique model to study renal development and disease at larger scale. Ideally, scoring of pronephric phenotypes includes morphological and functional assessments within the same larva. However, to efficiently upscale such assays, refinement of existing methods is required. Here, we describe the development of a multiparametric in vivo screening pipeline for parallel assessment of pronephric morphology, kidney function and heart rate within the same larva on a single imaging platform. To this end, we developed a novel 3D-printed orientation tool enabling multiple consistent orientations of larvae in agarose-filled microplates. Dorsal pronephros imaging was followed by assessing renal clearance and heart rates upon fluorescein isothiocyanate (FITC)-inulin microinjection using automated time-lapse imaging of laterally positioned larvae. The pipeline was benchmarked using a set of drugs known to induce developmental nephrotoxicity in humans and zebrafish. Drug-induced reductions in renal clearance and heart rate alterations were detected even in larvae exhibiting minor pronephric phenotypes. In conclusion, the developed workflow enables rapid and semi-automated in vivo assessment of multiple morphological and functional parameters.


Subject(s)
Biological Assay/methods , Heart Function Tests , Heart Rate/physiology , Kidney/physiology , Pronephros/anatomy & histology , Zebrafish/anatomy & histology , Zebrafish/physiology , Animals , Embryo, Nonmammalian/physiology , Fluorescein-5-isothiocyanate/metabolism , Larva/physiology , Pronephros/embryology , Zebrafish/embryology
6.
Cell Cycle ; 18(20): 2683-2696, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31451030

ABSTRACT

Zebrafish erythropoietin a (epoa) is a well characterized regulator of red blood cell formation. Recent morpholino mediated knockdown data have also identified epoa being essential for physiological pronephros development in zebrafish, which is driven by blocking apoptosis in developing kidneys. Yet, zebrafish mutants for epoa have not been described so far. In order to compare a transient knockdown vs. permanent knockout for epoa in zebrafish on pronephros development, we used CRISPR/Cas9 technology to generate epoa knockout zebrafish mutants and we performed structural and functional studies on pronephros development. In contrast to epoa morphants, epoa-/- zebrafish mutants showed normal pronephros structure; however, a previously uncharacterized gene in zebrafish, named epob, was identified and upregulated in epoa-/- mutants. epob knockdown altered pronephros development, which was further aggravated in epoa-/- mutants. Likewise, epoa and epob morphants regulated similar and differential gene signatures related to kidney development in zebrafish. In conclusion, stable loss of epoa during embryonic development can be compensated by epob leading to phenotypical discrepancies in epoa knockdown and knockout zebrafish embryos.


Subject(s)
Erythropoietin/metabolism , Gene Expression Regulation, Developmental/genetics , Organogenesis/genetics , Pronephros/embryology , Zebrafish Proteins/metabolism , Zebrafish/genetics , Animals , CRISPR-Cas Systems , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/ultrastructure , Erythropoietin/genetics , Gene Knockdown Techniques , Gene Knockout Techniques , Heterozygote , Homozygote , Microscopy, Electron , Morpholinos/genetics , Pronephros/abnormalities , Pronephros/metabolism , Recombinant Proteins/genetics , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/genetics
7.
Development ; 146(13)2019 07 10.
Article in English | MEDLINE | ID: mdl-31160420

ABSTRACT

Renal functional units known as nephrons undergo patterning events during development that create a segmental array of cellular compartments with discrete physiological identities. Here, from a forward genetic screen using zebrafish, we report the discovery that transcription factor AP-2 alpha (tfap2a) coordinates a gene regulatory network that activates the terminal differentiation program of distal segments in the pronephros. We found that tfap2a acts downstream of Iroquois homeobox 3b (irx3b), a distal lineage transcription factor, to operate a circuit consisting of tfap2b, irx1a and genes encoding solute transporters that dictate the specialized metabolic functions of distal nephron segments. Interestingly, this regulatory node is distinct from other checkpoints of differentiation, such as polarity establishment and ciliogenesis. Thus, our studies reveal insights into the genetic control of differentiation, where tfap2a is essential for regulating a suite of segment transporter traits at the final tier of zebrafish pronephros ontogeny. These findings have relevance for understanding renal birth defects, as well as efforts to recapitulate nephrogenesis in vivo to facilitate drug discovery and regenerative therapies.


Subject(s)
Kidney/embryology , Nephrons/embryology , Organogenesis/genetics , Transcription Factor AP-2/physiology , Zebrafish Proteins/physiology , Animals , Animals, Genetically Modified , Body Patterning/genetics , Cell Differentiation/genetics , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , Genes, Switch/physiology , Kidney/metabolism , Nephrons/metabolism , Pronephros/embryology , Pronephros/growth & development , Pronephros/metabolism , Transcription Factor AP-2/genetics , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
8.
Sci Rep ; 9(1): 6454, 2019 04 23.
Article in English | MEDLINE | ID: mdl-31015532

ABSTRACT

The genetic regulation of nephron patterning during kidney organogenesis remains poorly understood. Nephron tubules in zebrafish are composed of segment populations that have unique absorptive and secretory roles, as well as multiciliated cells (MCCs) that govern fluid flow. Here, we report that the transcription factor iroquois 2a (irx2a) is requisite for zebrafish nephrogenesis. irx2a transcripts localized to the developing pronephros and maturing MCCs, and loss of function altered formation of two segment populations and reduced MCC number. Interestingly, irx2a deficient embryos had reduced expression of an essential MCC gene ets variant 5a (etv5a), and were rescued by etv5a overexpression, supporting the conclusion that etv5a acts downstream of irx2a to control MCC ontogeny. Finally, we found that retinoic acid (RA) signaling affects the irx2a expression domain in renal progenitors, positioning irx2a downstream of RA. In sum, this work reveals new roles for irx2a during nephrogenesis, identifying irx2a as a crucial connection between RA signaling, segmentation, and the control of etv5a mediated MCC formation. Further investigation of the genetic players involved in these events will enhance our understanding of the molecular pathways that govern renal development, which can be used help create therapeutics to treat congenital and acquired kidney diseases.


Subject(s)
Cell Differentiation , Organogenesis/physiology , Pronephros/embryology , Transcription Factors/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Transcription Factors/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
9.
Integr Comp Biol ; 59(1): 29-47, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30649320

ABSTRACT

Contemporary papers and book chapters on nephrology open with the assumption that human kidney development passes through three morphological stages: pronephros, mesonephros, and metanephros. Current knowledge of the human pronephros, however, appears to be based on only a hand full of human specimens. The ongoing use of variations in the definition of a pronephros hampers the interpretation of study results. Because of the increased interest in the anamniote pronephros as a genetic model for kidney organogenesis we aimed to provide an overview of the literature concerning kidney development and to clarify the existence of a pronephros in human embryos. We performed an extensive literature survey regarding vertebrate renal morphology and we investigated histological sections of human embryos between 2 and 8 weeks of development. To facilitate better understanding of the literature about kidney development, a referenced glossary with short definitions was composed. The most striking difference between pronephros versus meso- and metanephros is found in nephron architecture. The pronephros consists exclusively of non-integrated nephrons with external glomeruli, whereas meso- and metanephros are composed of integrated nephrons with internal glomeruli. Animals whose embryos have comparatively little yolk at their disposal and hence have a free-swimming larval stage do develop a pronephros that is dedicated to survival in aquatic environments. Species in which embryos do not have a free-swimming larval stage have embryos that are supplied with a large amount of yolk or that develop within the body of the parent. In those species the pronephros is usually absent, incompletely developed, and apparently functionless. Non-integrated nephrons were not identified in histological sections of human embryos. Therefore, we conclude that a true pronephros is not detectable in human embryos although the most cranial part of the amniote excretory organ is often confusingly referred to as pronephros. The term pronephros should be avoided in amniotes unless all elements for a functional pronephros are undeniably present.


Subject(s)
Kidney/embryology , Pronephros/embryology , Vertebrates/embryology , Animals , Humans , Kidney/anatomy & histology , Pronephros/anatomy & histology , Vertebrates/anatomy & histology
10.
Sci Rep ; 8(1): 18038, 2018 12 21.
Article in English | MEDLINE | ID: mdl-30575756

ABSTRACT

Vertebrate kidneys contain nephron functional units where specialized epithelial cell types are organized into segments with discrete physiological roles. Many gaps remain in our understanding of how segment regions develop. Here, we report that the transcription factor empty spiracles homeobox gene 1 (emx1) is a novel nephron segment regulator during embryonic kidney development in zebrafish. emx1 loss of function altered the domains of distal segments without changes in cell turnover or traits like size and morphology, indicating that emx1 directs distal segment fates during nephrogenesis. In exploring how emx1 influences nephron patterning, we found that retinoic acid (RA), a morphogen that induces proximal and represses distal segments, negatively regulates emx1 expression. Next, through a series of genetic studies, we found that emx1 acts downstream of a cascade involving mecom and tbx2b, which encode essential distal segment transcription factors. Finally, we determined that emx1 regulates the expression domains of irx3b and irx1a to control distal segmentation, and sim1a to control corpuscle of Stannius formation. Taken together, our work reveals for the first time that emx1 is a key component of the pronephros segmentation network, which has implications for understanding the genetic regulatory cascades that orchestrate vertebrate nephron patterning.


Subject(s)
Homeodomain Proteins/physiology , Kidney/embryology , Nephrons/embryology , Organogenesis/genetics , Transcription Factors/physiology , Zebrafish , Animals , Animals, Genetically Modified , Body Patterning/genetics , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Kidney/metabolism , Nephrons/metabolism , Pronephros/embryology , Pronephros/metabolism , Transcription Factors/genetics , Zebrafish/embryology , Zebrafish/genetics
11.
Elife ; 72018 11 26.
Article in English | MEDLINE | ID: mdl-30475208

ABSTRACT

Nephron segmentation involves a concert of genetic and molecular signals that are not fully understood. Through a chemical screen, we discovered that alteration of peroxisome proliferator-activated receptor (PPAR) signaling disrupts nephron segmentation in the zebrafish embryonic kidney (Poureetezadi et al., 2016). Here, we show that the PPAR co-activator ppargc1a directs renal progenitor fate. ppargc1a mutants form a small distal late (DL) segment and an expanded proximal straight tubule (PST) segment. ppargc1a promotes DL fate by regulating the transcription factor tbx2b, and restricts expression of the transcription factor sim1a to inhibit PST fate. Interestingly, sim1a restricts ppargc1a expression to promote the PST, and PST development is fully restored in ppargc1a/sim1a-deficient embryos, suggesting Ppargc1a and Sim1a counterbalance each other in an antagonistic fashion to delineate the PST segment boundary during nephrogenesis. Taken together, our data reveal new roles for Ppargc1a during development, which have implications for understanding renal birth defects.


Subject(s)
Body Patterning , Nephrons/embryology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Amino Acid Sequence , Animals , Bezafibrate/pharmacology , Body Patterning/drug effects , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental/drug effects , Genetic Testing , Morpholinos/pharmacology , Nephrons/drug effects , Nephrons/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/chemistry , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Phenotype , Pronephros/drug effects , Pronephros/embryology , Pronephros/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Small Molecule Libraries/pharmacology , Transcription Factors/metabolism , Zebrafish/genetics , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics
12.
Microsc Res Tech ; 81(11): 1286-1294, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30341968

ABSTRACT

The embryonic origin of the urogenital system came from the intermediate mesoderm. Kidney development involves three successive renal systems with a fast chronological overlap: the pronephro, the mesonephro, and the metanephro. Due to the lack of specific knowledge about this system in cats the present work aimed to describe their urinary organs development, focusing on the structures seen in pronephro, mesonephro, and metanephro during the embryonic and fetal stages of development. The techniques used in this study were: light microscopy, immunohistochemistry, scanning electron microscopy, and transmission electron microscopy. For that, embryos and fetuses from 12 pregnant mixed-breed domestic cats in different gestational stages were used to describe the proposed organs. The pronephro is present at early stages of embryonary development in embryos from 15 to 19 days with the presence of pronephro's corpuscles, ducts and tubules. The mesonephro is found, in general, between days 17 and 37, and contains mesonephric ducts, mesonephric tubules, and glomeruli. The metanephro is seen since 21 days of pregnancy with the presence of glomeruli, proximal and distal contorted tubules and at day 37, the cortex-medullary region is already differentiated. The evaluation of these structures enhances the knowledge about embryology of the urinary system in cats, aiding a better anatomical understanding of the system in the specie allowing the correlation with other species.


Subject(s)
Embryo, Mammalian/embryology , Embryonic Development/physiology , Kidney Glomerulus/embryology , Mesonephros/embryology , Pronephros/embryology , Animals , Cats , Female , Immunohistochemistry , Kidney Glomerulus/anatomy & histology , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Pregnancy
13.
Biochem Biophys Res Commun ; 495(2): 1580-1587, 2018 01 08.
Article in English | MEDLINE | ID: mdl-29223398

ABSTRACT

Eph/ephrin molecules are widely expressed during embryonic development, and function in a variety of developmental processes. Here we studied the roles of the Eph receptor EphA7 and its soluble form in Xenopus pronephros development. EphA7 is specifically expressed in pronephric tubules at tadpole stages and knockdown of EphA7 by a translation blocking morpholino led to defects in tubule cell differentiation and morphogenesis. A soluble form of EphA7 (sEphA7) was also identified. Interestingly, the membrane level of claudin6 (CLDN6), a tetraspan transmembrane tight junction protein, was dramatically reduced in the translation blocking morpholino injected embryos, but not when a splicing morpholino was used, which blocks only the full length EphA7. In cultured cells, EphA7 binds and phosphorylates CLDN6, and reduces its distribution at the cell surface. Our work suggests a role of EphA7 in the regulation of cell adhesion during pronephros development, whereas sEphA7 works as an antagonist.


Subject(s)
Claudins/metabolism , Pronephros/embryology , Receptor, EphA7/metabolism , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Xenopus laevis/metabolism , Animals , Cell Membrane/metabolism , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Oligodeoxyribonucleotides, Antisense/genetics , Pronephros/metabolism , Receptor, EphA7/antagonists & inhibitors , Receptor, EphA7/genetics , Solubility , Xenopus Proteins/antagonists & inhibitors , Xenopus Proteins/genetics , Xenopus laevis/genetics
14.
Sci Rep ; 7(1): 8874, 2017 08 21.
Article in English | MEDLINE | ID: mdl-28827763

ABSTRACT

Peroxiredoxin1 (Prdx1) is an antioxidant enzyme belonging to the peroxiredoxin family of proteins. Prdx1 catalyzes the reduction of H2O2 and alkyl hydroperoxide and plays an important role in different biological processes. Prdx1 also participates in various age-related diseases and cancers. In this study, we investigated the role of Prdx1 in pronephros development during embryogenesis. Prdx1 knockdown markedly inhibited proximal tubule formation in the pronephros and significantly increased the cellular levels of reactive oxygen species (ROS), which impaired primary cilia formation. Additionally, treatment with ROS (H2O2) severely disrupted proximal tubule formation, whereas Prdx1 overexpression reversed the ROS-mediated inhibition in proximal tubule formation. Epistatic analysis revealed that Prdx1 has a crucial role in retinoic acid and Wnt signaling pathways during pronephrogenesis. In conclusion, Prdx1 facilitates proximal tubule formation during pronephrogenesis by regulating ROS levels.


Subject(s)
Peroxiredoxins/metabolism , Pronephros/embryology , Pronephros/metabolism , Reactive Oxygen Species/metabolism , Tretinoin/metabolism , Wnt Signaling Pathway , Amino Acid Sequence , Animals , Conserved Sequence , Cysteine , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Organogenesis/genetics , Peroxiredoxins/chemistry , Peroxiredoxins/genetics , Phenotype , Xenopus laevis
15.
Dev Biol ; 428(1): 148-163, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28579318

ABSTRACT

The zebrafish kidney is conserved with other vertebrates, making it an excellent genetic model to study renal development. The kidney collects metabolic waste using a blood filter with specialized epithelial cells known as podocytes. Podocyte formation is poorly understood but relevant to many kidney diseases, as podocyte injury leads to progressive scarring and organ failure. zeppelin (zep) was isolated in a forward screen for kidney mutants and identified as a homozygous recessive lethal allele that causes reduced podocyte numbers, deficient filtration, and fluid imbalance. Interestingly, zep mutants had a larger interrenal gland, the teleostean counterpart of the mammalian adrenal gland, which suggested a fate switch with the related podocyte lineage since cell proliferation and cell death were unchanged within the shared progenitor field from which these two identities arise. Cloning of zep by whole genome sequencing (WGS) identified a splicing mutation in breast cancer 2, early onset (brca2)/fancd1, which was confirmed by sequencing of individual fish. Several independent brca2 morpholinos (MOs) phenocopied zep, causing edema, reduced podocyte number, and increased interrenal cell number. Complementation analysis between zep and brca2ZM_00057434 -/- zebrafish, which have an insertional mutation, revealed that the interrenal lineage was expanded. Importantly, overexpression of brca2 rescued podocyte formation in zep mutants, providing critical evidence that the brca2 lesion encoded by zep specifically disrupts the balance of nephrogenesis. Taken together, these data suggest for the first time that brca2/fancd1 is essential for vertebrate kidney ontogeny. Thus, our findings impart novel insights into the genetic components that impact renal development, and because BRCA2/FANCD1 mutations in humans cause Fanconi anemia and several common cancers, this work has identified a new zebrafish model to further study brca2/fancd1 in disease.


Subject(s)
BRCA2 Protein/genetics , Gene Expression Regulation, Developmental , Organogenesis/genetics , Podocytes/cytology , Pronephros/embryology , Zebrafish Proteins/genetics , Zebrafish/embryology , Animals , Animals, Genetically Modified , Cell Differentiation , Cell Proliferation , Cloning, Molecular , Disease Models, Animal , In Situ Hybridization, Fluorescence , Morpholinos/genetics , Pronephros/cytology , Zebrafish/genetics
16.
Dev Biol ; 425(2): 130-141, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28359809

ABSTRACT

During zebrafish embryogenesis the pronephric kidney arises from a small population of posterior mesoderm cells that then undergo expansion during early stages of renal organogenesis. While wnt8 is required for posterior mesoderm formation during gastrulation, it is also transiently expressed in the post-gastrula embryo in the intermediate mesoderm, the precursor to the pronephros and some blood/vascular lineages. Here, we show that knockdown of wnt8a, using a low dose of morpholino that does not disrupt early mesoderm patterning, reduces the number of kidney and blood cells. For the kidney, wnt8a deficiency decreases renal progenitor growth during early somitogenesis, as detected by EdU incorporation, but has no effect on apoptosis. The depletion of the renal progenitor pool in wnt8a knockdown embryos leads to cellular deficits in the pronephros at 24 hpf that are characterised by a shortened distal-most segment and stretched proximal tubule cells. A pulse of the canonical Wnt pathway agonist BIO during early somitogenesis is sufficient to rescue the size of the renal progenitor pool while longer treatment expands the number of kidney cells. Taken together, these observations indicate that Wnt8, in addition to its well-established role in posterior mesoderm patterning, also plays a later role as a factor that expands the renal progenitor pool prior to kidney morphogenesis.


Subject(s)
Cytoskeletal Proteins/metabolism , Embryo, Nonmammalian/cytology , Kidney/cytology , Kidney/embryology , Stem Cells/cytology , Wnt Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Animals , Apoptosis/drug effects , Blood Cells/cytology , Blood Cells/drug effects , Body Patterning/drug effects , Cell Count , Cell Proliferation/drug effects , Embryo, Nonmammalian/drug effects , Gene Knockdown Techniques , Indoles/pharmacology , Kidney Tubules/drug effects , Kidney Tubules/pathology , Mesoderm/drug effects , Mesoderm/embryology , Mesoderm/metabolism , Morpholinos/pharmacology , Oximes/pharmacology , Pronephros/cytology , Pronephros/embryology , Stem Cells/drug effects , Stem Cells/metabolism
17.
Results Probl Cell Differ ; 60: 27-53, 2017.
Article in English | MEDLINE | ID: mdl-28409341

ABSTRACT

The pronephros is the first kidney type to form in vertebrate embryos. The first step of pronephrogenesis in the zebrafish is the formation of the intermediate mesoderm during gastrulation, which occurs in response to secreted morphogens such as BMPs and Nodals. Patterning of the intermediate mesoderm into proximal and distal cell fates is induced by retinoic acid signaling with downstream transcription factors including wt1a, pax2a, pax8, hnf1b, sim1a, mecom, and irx3b. In the anterior intermediate mesoderm, progenitors of the glomerular blood filter migrate and fuse at the midline and recruit a blood supply. More posteriorly localized tubule progenitors undergo epithelialization and fuse with the cloaca. The Notch signaling pathway regulates the formation of multi-ciliated cells in the tubules and these cells help propel the filtrate to the cloaca. The lumenal sheer stress caused by flow down the tubule activates anterior collective migration of the proximal tubules and induces stretching and proliferation of the more distal segments. Ultimately these processes create a simple two-nephron kidney that is capable of reabsorbing and secreting solutes and expelling excess water-processes that are critical to the homeostasis of the body fluids. The zebrafish pronephric kidney provides a simple, yet powerful, model system to better understand the conserved molecular and cellular progresses that drive nephron formation, structure, and function.


Subject(s)
Pronephros/embryology , Zebrafish/embryology , Animals , Models, Animal , Organogenesis , Zebrafish Proteins
18.
Biochem Biophys Res Commun ; 487(2): 209-215, 2017 May 27.
Article in English | MEDLINE | ID: mdl-28411024

ABSTRACT

Epithelial cell adhesion molecule EpCAM is a transmembrane glycoprotein that is dynamically expressed in human and murine renal epithelia during development. The levels of EpCAM in the renal epithelium are upregulated both during regeneration after ischemia/reperfusion injury and in renal-derived carcinomas. The role of EpCAM in early kidney development, however, has remained unclear. The zebrafish pronephros shows a similar segmentation pattern to the mammalian metanephric nephron, and has recently emerged as a tractable model to study the regulatory programs governing early nephrogenesis. Since EpCAM shows persistent expression in the pronephros throughout early development, we developed a method to study the global changes in gene expression in specific pronephric segments of wild type and EpCAM-deficient zebrafish embryos. In epcam mutants, we found 379 differentially expressed genes. Gene ontology analysis revealed that EpCAM controls various developmental programs, including uretric bud development, morphogenesis of branching epithelium, regulation of cell differentiation and cilium morphogenesis.


Subject(s)
Membrane Glycoproteins/metabolism , Morphogenesis/physiology , Pronephros/embryology , Pronephros/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Animals , Gene Expression Regulation, Developmental/physiology , Pronephros/growth & development , Zebrafish/growth & development
19.
Dev Biol ; 421(1): 52-66, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27840199

ABSTRACT

The simplified and genetically conserved zebrafish pronephros is an excellent model to examine the cryptic processes of cell fate decisions during the development of nephron segments as well as the origins of associated endocrine cells that comprise the corpuscles of Stannius (CS). Using whole mount in situ hybridization, we found that transcripts of the zebrafish genes t-box 2a (tbx2a) and t-box 2b (tbx2b), which belong to the T-box family of transcription factors, were expressed in the caudal intermediate mesoderm progenitors that give rise to the distal pronephros and CS. Deficiency of tbx2a, tbx2b or both tbx2a/b reduced the size of the distal late (DL) segment, which was accompanied by a proximal convoluted segment (PCT) expansion. Further, tbx2a/b deficiency led to significantly larger CS clusters. These phenotypes were also observed in embryos with the from beyond (fby)c144 mutation, which encodes a premature stop codon in the tbx2b T-box sequence. Conversely, overexpression of tbx2a and tbx2b in wild-type embryos expanded the DL segment where cells were comingled with the adjacent DE, and also decreased CS cell number, but notably did not alter PCT development-providing independent evidence that tbx2a and tbx2b are each necessary and sufficient to promote DL fate and suppress CS genesis. Epistasis studies indicated that tbx2a acts upstream of tbx2b to regulate the DL and CS fates, and likely has other targets as well. Retinoic acid (RA) addition and inhibition studies revealed that tbx2a and tbx2b are negatively regulated by RA signaling. Interestingly, the CS cell expansion that typifies tbx2a/b deficiency also occurred when blocking Notch signaling with the chemical DAPT (N-[N-(3,5-difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester). Ectopic activation of Notch in Tg(hsp70::Gal4; UAS::NICD)(NICD) embryos led to a reduced CS post heat-shock induction. To further examine the link between the tbx2a/b genes and Notch during CS formation, DAPT treatment was used to block Notch activity in tbx2a/b deficient embryos, and tbx2a/b knockdown was performed in NICD transgenic embryos. Both manipulations caused similar CS expansions, indicating that Notch functions upstream of the tbx2a/b genes to suppress CS ontogeny. Taken together, these data reveal for the first time that tbx2a/b mitigate pronephros segmentation downstream of RA, and that interplay between Notch signaling and tbx2a/b regulate CS formation, thus providing several novel insights into the genetic regulatory networks that influence these lineages.


Subject(s)
Body Patterning/genetics , Pronephros/embryology , Pronephros/metabolism , T-Box Domain Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Animals , Body Patterning/drug effects , Cell Count , Cell Differentiation/drug effects , Gene Expression Regulation, Developmental/drug effects , Mesoderm/drug effects , Mesoderm/embryology , Mesoderm/metabolism , Models, Biological , Organogenesis/drug effects , Organogenesis/genetics , Pronephros/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Notch/metabolism , Signal Transduction/drug effects , T-Box Domain Proteins/genetics , Tretinoin/pharmacology , Zebrafish/genetics , Zebrafish Proteins/genetics
20.
Kidney Blood Press Res ; 41(6): 794-801, 2016.
Article in English | MEDLINE | ID: mdl-27832650

ABSTRACT

BACKGROUNDS/AIMS: Vitamin C is an antioxidant and acts as a cofactor for several key enzymatic catalytic reactions in animals. Amphibians produce vitamin C in their kidneys, as opposed to mammals that produce vitamin C in their liver. Gulo serves as a crucial enzyme for vitamin C synthesis in mammals, but the characteristics and localization of its homologous genes during kidney development in Xenopus laevis, an amphibian, remains unknown. METHODS: We aligned amino acid sequences of Gulo across different species by using bioinformatics methods and detected patterns of expression for Gulo during kidney development by using RT-PCR and in situ hybridization. RESULTS: We identified a new site on the X. laevis genome, LOC495407. Sequence alignment analysis indicated this fragment is highly conserved and homologous to gulo genes in mammals. RT-PCR and in situ hybridization results reveal that X. laevis gulo is maternally expressed during the early stages of embryonic development, particularly, in the tubules of the pronephros from the middle tail-bud stage and onward in embryos. CONCLUSION: Gulo is a novel specific marker for pronephros tubules in X. laevis, and may be used as a potential marker for kidney development studies and disease diagnosis in mammals.


Subject(s)
Kidney Tubules/growth & development , L-Gulonolactone Oxidase/analysis , Pronephros/growth & development , Animals , Biomarkers/analysis , Female , Kidney Tubules/embryology , Kidney Tubules/enzymology , Mammals , Pronephros/embryology , Pronephros/enzymology , Sequence Alignment , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...